1
|
Xiao T, Cheng X, Zhi Y, Tian F, Wu A, Huang F, Tao L, Guo Z, Shen X. Ameliorative effect of Alangium chinense (Lour.) Harms on rheumatoid arthritis by reducing autophagy with targeting regulate JAK3-STAT3 and COX-2 pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117133. [PMID: 37690476 DOI: 10.1016/j.jep.2023.117133] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alangium chinense has been used as traditional folk medicine for centuries to treat rheumatoid arthritis (RA) by Guizhou Miao nationality with remarkable clinical effect. But the mechanism of its anti-RA is not fully clarified. AIM OF THE STUDY To explore the effect and underlying mechanism of A. chinense against RA. MATERIAL AND METHODS RA rats were induced by CII/IFA, and oral administrated with or without ethyl acetate extracts of Alangium chinense (ACEE) and tripterygium glycosides (GTW). Then arthritis scores, inflammatory factors in serum and histological evaluation were evaluated to assess the degree of joints disease. Proteomics were conducted via LC-MS/MS to clarify the mechanism of ACEE preliminarily, and further examined by immunohistochemistry, immunofluorescence, western botting, and molecular docking. RESULTS ACEE decreased joints swelling, cell abscission and necrosis of joint tissues arthropathy of RA rats, and attenuated expression of TNF-α, IL-1β, IL-6, PGE2, TGF-β. Meanwhile, differentially expressed proteins in the ACEE treated groups were observed, which were involved in RA, spliceosome, cell adhesion molecules, phagosome and lysosome signaling pathways. Moreover, ACEE significantly ameliorated arthropathy, suppressed JAK-STAT pathway (JAK3, p-JAK3, STAT3, iNOS, RANKL), COX-2 pathway (COX-2, TNF-α, IL-6I, L-1β, 5-LOX), and autophagic signaling pathway (LC3-Ⅰ, LC3-Ⅱ, p62, mTOR). But it showed little effect on the expression of COX-1, JAK1, JAK2, TyK2. CONCLUSION It is the first evidence that A. chinense significantly ameliorates RA, and the underlying immune mechanism involves reducing autophagy with targeting regulate JAK3-STAT3 and COX-2 pathways.
Collapse
Affiliation(s)
- Ting Xiao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Xingyan Cheng
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Yuan Zhi
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Fangfang Tian
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Ai Wu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Feilong Huang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Ling Tao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Zhenghong Guo
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| |
Collapse
|
2
|
Benziger PT, Kopping EJ, McLaughlin PA, Thanassi DG. Francisella tularensis disrupts TLR2-MYD88-p38 signaling early during infection to delay apoptosis of macrophages and promote virulence in the host. mBio 2023; 14:e0113623. [PMID: 37404047 PMCID: PMC10470500 DOI: 10.1128/mbio.01136-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 05/24/2023] [Indexed: 07/06/2023] Open
Abstract
Francisella tularensis is a zoonotic pathogen and the causative agent of tularemia. F. tularensis replicates to high levels within the cytosol of macrophages and other host cells while subverting the host response to infection. Critical to the success of F. tularensis is its ability to delay macrophage apoptosis to maintain its intracellular replicative niche. However, the host-signaling pathway(s) modulated by F. tularensis to delay apoptosis are poorly characterized. The outer membrane channel protein TolC is required for F. tularensis virulence and its ability to suppress apoptosis and cytokine expression during infection of macrophages. We took advantage of the F. tularensis ∆tolC mutant phenotype to identify host pathways that are important for activating macrophage apoptosis and that are disrupted by the bacteria. Comparison of macrophages infected with wild-type or ∆tolC F. tularensis revealed that the bacteria interfere with TLR2-MYD88-p38 signaling at early times post infection to delay apoptosis, dampen innate host responses, and preserve the intracellular replicative niche. Experiments using the mouse pneumonic tularemia model confirmed the in vivo relevance of these findings, revealing contributions of TLR2 and MYD88 signaling to the protective host response to F. tularensis, which is modulated by the bacteria to promote virulence. IMPORTANCE Francisella tularensis is a Gram-negative intracellular bacterial pathogen and the causative agent of the zoonotic disease tularemia. F. tularensis, like other intracellular pathogens, modulates host-programmed cell death pathways to ensure its replication and survival. We previously identified the outer membrane channel protein TolC as required for the ability of F. tularensis to delay host cell death. However, the mechanism by which F. tularensis delays cell death pathways during intracellular replication is unclear despite being critical to pathogenesis. In the present study, we address this gap in knowledge by taking advantage of ∆tolC mutants of F. tularensis to uncover signaling pathways governing host apoptotic responses to F. tularensis and which are modulated by the bacteria during infection to promote virulence. These findings reveal mechanisms by which intracellular pathogens subvert host responses and enhance our understanding of the pathogenesis of tularemia.
Collapse
Affiliation(s)
- P. Todd Benziger
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - Erik J. Kopping
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - Patrick A. McLaughlin
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - David G. Thanassi
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
3
|
Zhao Y, Yang Y, Liu M, Qin X, Yu X, Zhao H, Li X, Li W. COX-2 is required to mediate crosstalk of ROS-dependent activation of MAPK/NF-κB signaling with pro-inflammatory response and defense-related NO enhancement during challenge of macrophage-like cell line with Giardia duodenalis. PLoS Negl Trop Dis 2022; 16:e0010402. [PMID: 35482821 PMCID: PMC9089906 DOI: 10.1371/journal.pntd.0010402] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 05/10/2022] [Accepted: 04/08/2022] [Indexed: 01/07/2023] Open
Abstract
Giardia duodenalis, the causative agent of giardiasis, is among the most important causes of waterborne diarrheal diseases around the world. Giardia infection may persist over extended periods with intestinal inflammation, although minimal. Cyclooxygenase (COX)-2 is well known as an important inducer of inflammatory response, while the role it played in noninvasive Giardia infection remains elusive. Here we investigated the regulatory function of COX-2 in Giardia-induced pro-inflammatory response and defense-related nitric oxide (NO) generation in macrophage-like cell line, and identified the potential regulators. We initially found that Giardia challenge induced up-regulation of IL-1β, IL-6, TNF-α, prostaglandin (PG) E2, and COX-2 in macrophages, and pretreatment of the cells with COX-2 inhibitor NS398 reduced expressions of those pro-inflammatory factors. It was also observed that COX-2 inhibition could attenuate the up-regulated NO release and inducible NO synthase (iNOS) expression induced by Giardia. We further confirmed that Giardia-induced COX-2 up-regulation was mediated by the phosphorylation of p38 and ERK1/2 MAPKs and NF-κB. In addition, inhibition of reactive oxygen species (ROS) by NAC was shown to repress Giardia-induced activation of MAPK/NF-κB signaling, up-regulation of COX-2 and iNOS, increased levels of PGE2 and NO release, and up-expressions of IL-1β, IL-6, and TNF-α. Collectively, in this study, we revealed a critical role of COX-2 in modulating pro-inflammatory response and defense-related NO production in Giardia-macrophage interactions, and this process was evident to be controlled by ROS-dependent activation of MAPK/NF-κB signaling. The results can deepen our knowledge of anti-Giardia inflammatory response and host defense mechanisms.
Collapse
Affiliation(s)
- Yudan Zhao
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yongwu Yang
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Min Liu
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xuening Qin
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xiran Yu
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Huimin Zhao
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xiaoyun Li
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Wei Li
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- * E-mail:
| |
Collapse
|
4
|
Feng C, Bao X, Shan L, Ling Y, Ding Y, Wang J, Cao Y, Wang Q, Cui W, Xu S. Calcium-Sensing Receptor Mediates β-Amyloid-Induced Synaptic Formation Impairment and Cognitive Deficits via Regulation of Cytosolic Phospholipase A2/Prostaglandin E2 Metabolic Pathway. Front Aging Neurosci 2020; 12:144. [PMID: 32670047 PMCID: PMC7328130 DOI: 10.3389/fnagi.2020.00144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/29/2020] [Indexed: 12/15/2022] Open
Abstract
Calcium-sensing receptor (CaSR) is a G protein-coupled receptor (GPCRs). Soluble β-amyloid peptide (Aβ) is one of the orthosteric modulators of CaSR, while, the role and underlying mechanism of CaSR in cognitive decline in Alzheimer’s disease (AD) is unclear. In this study, molecular technology such as live-cell imaging combined with behavioral tests were used to explore the role and the underlying mechanism of CaSR in the cognitive deficits in AD mice. The expression levels of CaSR were increased both in AD mice and Aβ1–42 (β-amyloid protein)-treated primary cultured neurons. Pharmacological inhibition of CaSR ameliorated recognitive and spatial memory deficits of Aβ1–42-oligomer-treated mice in a dose-dependent manner. Pharmacological inhibition of CaSR or down-regulation of the expression of CaSR by CaSR-shRNA-lentivirus prevented the impairment of filopodia, and the synapse induced by oligomeric Aβ1–42. The contents of cytosolic phospholipase A2 (cPLA2) and prostaglandin E2 (PGE2) in hippocampal neurons and tissue were increased after treatment with Aβ1–42 oligomers. Inhibition or down-regulation of CaSR mediates Aβ-induced synapse formation and cognitive deficits partially, through the activation of the cPLA2/PGE2 pathway. This study provides novel insights on CaSR, which is a promising therapeutic target for AD.
Collapse
Affiliation(s)
- Chenxi Feng
- School of Medicine, Ningbo University, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo, China.,Children's Hospital of Soochow University, Suzhou, China
| | - Xiaoming Bao
- HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China.,Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Ling Shan
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Yunxiang Ling
- School of Medicine, Ningbo University, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo, China
| | - Yanfei Ding
- School of Medicine, Ningbo University, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo, China
| | - Jia Wang
- School of Medicine, Ningbo University, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo, China
| | - Yanzi Cao
- School of Medicine, Ningbo University, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo, China
| | - Qinwen Wang
- School of Medicine, Ningbo University, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo, China
| | - Wei Cui
- School of Medicine, Ningbo University, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo, China
| | - Shujun Xu
- School of Medicine, Ningbo University, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo, China
| |
Collapse
|
5
|
Yu G, Mo S, Gao L, Wen X, Chen S, Long X, Xie X, Deng Y, Ren L, Zang N, Chen S, Liu E. Club cell 10-kDa protein (CC10) inhibits cPLA2/COX2 pathway to alleviate RSV-induced airway inflammation and AHR. Int Immunopharmacol 2020; 83:106327. [DOI: 10.1016/j.intimp.2020.106327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/14/2020] [Accepted: 02/14/2020] [Indexed: 11/25/2022]
|
6
|
Novel Therapeutic Potentials of Taxifolin for Amyloid-β-associated Neurodegenerative Diseases and Other Diseases: Recent Advances and Future Perspectives. Int J Mol Sci 2019; 20:ijms20092139. [PMID: 31052203 PMCID: PMC6539020 DOI: 10.3390/ijms20092139] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 04/17/2019] [Accepted: 04/27/2019] [Indexed: 12/21/2022] Open
Abstract
Amyloid-β (Aβ) has been closely implicated in the pathogenesis of cerebral amyloid angiopathy (CAA) and Alzheimer’s disease (AD), the major causes of dementia. Thus, Aβ could be a target for the treatment of these diseases, for which, currently, there are no established effective treatments. Taxifolin is a bioactive catechol-type flavonoid present in various plants, such as herbs, and it exhibits pleiotropic effects including anti-oxidant and anti-glycation activities. Recently, we have demonstrated that taxifolin inhibits Aβ fibril formation in vitro and have further shown that it improves cerebral blood flow, facilitating Aβ clearance in the brain and suppressing cognitive decline in a mouse model of CAA. These findings suggest the novel therapeutic potentials of taxifolin for CAA. Furthermore, recent extensive studies have reported several novel aspects of taxifolin supporting its potential as a therapeutic drug for AD and metabolic diseases with a high risk for dementia as well as for CAA. In this review, we have summarized the recent advances in taxifolin research based on in vitro, in vivo, and in silico approaches. Furthermore, we have discussed future research directions on the potential of taxifolin for use in novel therapeutic strategies for CAA, AD, and metabolic diseases with an increased risk for dementia.
Collapse
|
7
|
Ford CL, Wang Y, Morgan K, Boktor M, Jordan P, Castor TP, Alexander JS. Interferon-gamma depresses human intestinal smooth muscle cell contractility: Relevance to inflammatory gut motility disturbances. Life Sci 2019; 222:69-77. [DOI: 10.1016/j.lfs.2019.01.059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 01/17/2019] [Accepted: 01/17/2019] [Indexed: 02/07/2023]
|
8
|
Kanikarla-Marie P, Kopetz S, Hawk ET, Millward SW, Sood AK, Gresele P, Overman M, Honn K, Menter DG. Bioactive lipid metabolism in platelet "first responder" and cancer biology. Cancer Metastasis Rev 2019; 37:439-454. [PMID: 30112590 DOI: 10.1007/s10555-018-9755-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Platelets can serve as "first responders" in cancer and metastasis. This is partly due to bioactive lipid metabolism that drives both platelet and cancer biology. The two primary eicosanoid metabolites that maintain platelet rapid response homeostasis are prostacyclin made by endothelial cells that inhibits platelet function, which is counterbalanced by thromboxane produced by platelets during activation, aggregation, and platelet recruitment. Both of these arachidonic acid metabolites are inherently unstable due to their chemical structure. Tumor cells by contrast predominantly make more chemically stable prostaglandin E2, which is the primary bioactive lipid associated with inflammation and oncogenesis. Pharmacological, clinical, and epidemiologic studies demonstrate that non-steroidal anti-inflammatory drugs (NSAIDs), which target cyclooxygenases, can help prevent cancer. Much of the molecular and biological impact of these drugs is generally accepted in the field. Cyclooxygenases catalyze the rate-limiting production of substrate used by all synthase molecules, including those that produce prostaglandins along with prostacyclin and thromboxane. Additional eicosanoid metabolites include lipoxygenases, leukotrienes, and resolvins that can also influence platelets, inflammation, and carcinogenesis. Our knowledge base and technology are now progressing toward identifying newer molecular and cellular interactions that are leading to revealing additional targets. This review endeavors to summarize new developments in the field.
Collapse
Affiliation(s)
- Preeti Kanikarla-Marie
- Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Scott Kopetz
- Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Ernest T Hawk
- Office of the Vice President Cancer Prevention and Population Science, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Steven W Millward
- Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Anil K Sood
- Gynocologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA.,Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA.,Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Paolo Gresele
- Department of Medicine, Section of Internal and Cardiovascular Medicine, University of Perugia, Via E. Dal Pozzo, 06126, Perugia, Italy
| | - Michael Overman
- Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Kenneth Honn
- Bioactive Lipids Research Program, Department of Pathology, Wayne State University, 5101 Cass Ave. 430 Chemistry, Detroit, MI, 48202, USA.,Department of Pathology, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA.,Cancer Biology Division, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA.,Department of Gastrointestinal Medical Oncology, M. D. Anderson Cancer Center, 1515 Holcombe Boulevard--Unit 0426, Houston, TX, 77030, USA
| | - David G Menter
- Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA.
| |
Collapse
|
9
|
Wang Y, Wang Q, Bao X, Ding Y, Shentu J, Cui W, Chen X, Wei X, Xu S. Taxifolin prevents β-amyloid-induced impairments of synaptic formation and deficits of memory via the inhibition of cytosolic phospholipase A 2/prostaglandin E 2 content. Metab Brain Dis 2018. [PMID: 29542038 DOI: 10.1007/s11011-018-0207-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Taxifolin is a potent flavonoid with anti-inflammatory activity. Taxifolin has been reported to decrease the accumulation of β-amyloid (Aβ), and reduce Aβ-induced neurotoxicity. However, the detail molecular mechanism of taxifolin against Aβ-induced neurotoxicity is largely unknown. In this study, we revealed the protective effects and the underlying mechanisms of taxifolin on the impairments of cognitive function and synapse formation induced by soluble Aβ oligomers. Our results showed that taxifolin prevented neuronal cell death in a concentration-dependent manner. The recognition memory in novel object recognition tasks and the spatial memory in Morris water maze tests are significantly lower in the Alzheimer's disease (AD) model mice induced by hippocampal injection of Aβ42. Taxifolin treatment prevented the recognitive and spatial memory deficits of the AD mice. 10 mg/kg taxifolin treatment also significantly prevented the decreased expression levels of PSD 95 induced by Aβ42. Live cell imaging study showed that 2 h pre-treatment of taxifolin prevented the decrease in the number of filopodium and spine induced by Aβ42 oligomers. Aβ42 oligomers significantly increased the production of cytosolic phospholipase A2 (cPLA2), a crucial enzyme of pro-inflammatory mediator, and prostaglandin E2 (PGE2), a neuroinflammatory molecule. Taxifolin significantly reduced the content of cPLA2 and PGE2 induced by Aβ42 both in the primary hippocampal neurons and hippocampal tissues. These results indicated that taxifolin might prevent Aβ42 oligomer-induced synapse and cognitive impairments through decreasing cPLA2 and PGE2. Our study provided novel insights into the cellular mechanisms for the protective effects of taxifolin on AD.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Qinwen Wang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Xiaoming Bao
- The No.2 Hospital of Ningbo, Ningbo, Zhejiang, 315010, China
| | - Yanfei Ding
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Jieyi Shentu
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Wei Cui
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Xiaowei Chen
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Xiaofei Wei
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Shujun Xu
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine Ningbo University, Ningbo, Zhejiang, 315211, China.
| |
Collapse
|
10
|
Fabrik I, Link M, Putzova D, Plzakova L, Lubovska Z, Philimonenko V, Pavkova I, Rehulka P, Krocova Z, Hozak P, Santic M, Stulik J. The Early Dendritic Cell Signaling Induced by Virulent Francisella tularensis Strain Occurs in Phases and Involves the Activation of Extracellular Signal-Regulated Kinases (ERKs) and p38 In the Later Stage. Mol Cell Proteomics 2017; 17:81-94. [PMID: 29046388 DOI: 10.1074/mcp.ra117.000160] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/22/2017] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells (DCs) infected by Francisella tularensis are poorly activated and do not undergo classical maturation process. Although reasons of such unresponsiveness are not fully understood, their impact on the priming of immunity is well appreciated. Previous attempts to explain the behavior of Francisella-infected DCs were hypothesis-driven and focused on events at later stages of infection. Here, we took an alternative unbiased approach by applying methods of global phosphoproteomics to analyze the dynamics of cell signaling in primary DCs during the first hour of infection by Francisella tularensis Presented results show that the early response of DCs to Francisella occurs in phases and that ERK and p38 signaling modules induced at the later stage are differentially regulated by virulent and attenuated ΔdsbA strain. These findings imply that the temporal orchestration of host proinflammatory pathways represents the integral part of Francisella life-cycle inside hijacked DCs.
Collapse
Affiliation(s)
- Ivo Fabrik
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Marek Link
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Daniela Putzova
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Lenka Plzakova
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Zuzana Lubovska
- §Institute of Molecular Genetics ASCR v.v.i., Microscopy Centre, Electron Microscopy Core Facility, 142 20 Prague 4, Czech Republic
| | - Vlada Philimonenko
- §Institute of Molecular Genetics ASCR v.v.i., Microscopy Centre, Electron Microscopy Core Facility, 142 20 Prague 4, Czech Republic.,¶Institute of Molecular Genetics ASCR v.v.i., Department of Biology of the Cell Nucleus, 142 20 Prague 4, Czech Republic
| | - Ivona Pavkova
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Pavel Rehulka
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Zuzana Krocova
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Pavel Hozak
- §Institute of Molecular Genetics ASCR v.v.i., Microscopy Centre, Electron Microscopy Core Facility, 142 20 Prague 4, Czech Republic.,¶Institute of Molecular Genetics ASCR v.v.i., Department of Biology of the Cell Nucleus, 142 20 Prague 4, Czech Republic
| | - Marina Santic
- ‖Department of Microbiology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Jiri Stulik
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic;
| |
Collapse
|
11
|
DeMars KM, Pacheco SC, Yang C, Siwarski DM, Candelario-Jalil E. Selective Inhibition of Janus Kinase 3 Has No Impact on Infarct Size or Neurobehavioral Outcomes in Permanent Ischemic Stroke in Mice. Front Neurol 2017; 8:363. [PMID: 28790974 PMCID: PMC5524742 DOI: 10.3389/fneur.2017.00363] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/10/2017] [Indexed: 11/20/2022] Open
Abstract
Janus kinase 3 (JAK3) is associated with the common gamma chain of several interleukin (IL) receptors essential to inflammatory signaling. To study the potential role of JAK3 in stroke-induced neuroinflammation, we subjected mice to permanent middle cerebral artery occlusion and investigated the effects of JAK3 inhibition with decernotinib (VX-509) on infarct size, behavior, and levels of several inflammatory mediators. Results from our double immunofluorescence staining showed JAK3 expression on neurons, endothelial cells, and microglia/macrophages in the ischemic mouse brain (n = 3). We found for the first time that total and phosphorylated/activated JAK3 are dramatically increased after stroke in the ipsilateral hemisphere (**P < 0.01; n = 5–13/group) in addition to increased IL-21 expression after stroke (**P < 0.01; n = 5–7/group). However, inhibition of JAK3 confirmed by reduced phosphorylation of its activation loop at tyrosine residues 980/981 does not reduce infarct volume measured at 48 h after stroke (n = 6–10/group) nor does it alter behavioral outcomes sensitive to neurological deficits or stroke-induced neuroinflammatory response (n = 9–10/group). These results do not support a detrimental role for JAK3 in acute neuroinflammation following permanent focal cerebral ischemia. The functional role of increased JAK3 activation after stroke remains to be further investigated.
Collapse
Affiliation(s)
- Kelly M DeMars
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Sean C Pacheco
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Changjun Yang
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - David M Siwarski
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
12
|
Ndiaye K, Castonguay A, Benoit G, Silversides DW, Lussier JG. Differential regulation of Janus kinase 3 (JAK3) in bovine preovulatory follicles and identification of JAK3 interacting proteins in granulosa cells. J Ovarian Res 2016; 9:71. [PMID: 27793176 PMCID: PMC5086056 DOI: 10.1186/s13048-016-0280-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/17/2016] [Indexed: 11/11/2022] Open
Abstract
Background Janus kinase 3 (JAK3) is a member of the membrane-associated non-receptor tyrosine kinase protein family and is considered predominantly expressed in hematopoietic cells. We previously identified JAK3 as a differentially expressed gene in granulosa cells (GC) of bovine preovulatory follicles. The present study aimed to further investigate JAK3 regulation, to identify protein binding partners and better understand its mode of action in bovine reproductive cells. Results GC were obtained from small follicles (SF), dominant follicles at day 5 of the estrous cycle (DF), and ovulatory follicles, 24 h following hCG injection (OF). RT-PCR analyses showed greatest expression of JAK3 in GC of DF, while JAK3 expression was downregulated in OF (P < 0.0001). In addition, there was a 5- and 20-fold reduction of JAK3 steady-state mRNA levels in follicular walls, respectively at 12 and 24 hours post-hCG as compared to 0 h (P < 0.05). Similarly, JAK3 expression was downregulated by the endogenous LH surge. These results were confirmed in western blot analysis showing weakest JAK3 protein amounts in OF as compared to DF. Yeast two-hybrid screening of a DF-cDNA library resulted in the identification of JAK3 partners in GC that were confirmed by co-immunoprecipitation and included leptin receptor overlapping transcript-like 1 (LEPROTL1), inhibin beta A (INHBA) and cyclin-dependent kinase inhibitor 1B (CDKN1B). In functional studies using bovine endometrial cells, JAK3 increased phosphorylation of STAT3 and cell viability, while the addition of JANEX-1 inhibited JAK3 actions. Conclusion These results support a physiologically relevant role of JAK3 in follicular development and provide insights into the mode of action and function of JAK3 in reproductive tissues. Electronic supplementary material The online version of this article (doi:10.1186/s13048-016-0280-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kalidou Ndiaye
- Département de biomédecine vétérinaire, Faculté de médecine vétérinaire, Centre de recherche en reproduction animale (CRRA), Université de Montréal, P.O. Box 5000, St-Hyacinthe, Québec, J2S 7C6, Canada. .,Faculté de médecine vétérinaire, Département de biomédecine vétérinaire, Université de Montréal, 3200 Rue Sicotte, St-Hyacinthe, Québec, J2S 2M2, Canada.
| | - Amélie Castonguay
- Département de biomédecine vétérinaire, Faculté de médecine vétérinaire, Centre de recherche en reproduction animale (CRRA), Université de Montréal, P.O. Box 5000, St-Hyacinthe, Québec, J2S 7C6, Canada
| | - Gabriel Benoit
- Département de biomédecine vétérinaire, Faculté de médecine vétérinaire, Centre de recherche en reproduction animale (CRRA), Université de Montréal, P.O. Box 5000, St-Hyacinthe, Québec, J2S 7C6, Canada
| | - David W Silversides
- Département de biomédecine vétérinaire, Faculté de médecine vétérinaire, Centre de recherche en reproduction animale (CRRA), Université de Montréal, P.O. Box 5000, St-Hyacinthe, Québec, J2S 7C6, Canada
| | - Jacques G Lussier
- Département de biomédecine vétérinaire, Faculté de médecine vétérinaire, Centre de recherche en reproduction animale (CRRA), Université de Montréal, P.O. Box 5000, St-Hyacinthe, Québec, J2S 7C6, Canada
| |
Collapse
|
13
|
Abstract
Various clinical and epidemiologic studies show that nonsteroidal anti-inflammatory drugs (NSAIDs), including aspirin and cyclooxygenase inhibitors (COXIBs) help prevent cancer. Since eicosanoid metabolism is the main inhibitory targets of these drugs the resulting molecular and biological impact is generally accepted. As our knowledge base and technology progress we are learning that additional targets may be involved. This review attempts to summarize these new developments in the field.
Collapse
Affiliation(s)
- Asad Umar
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Vernon E Steele
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - David G Menter
- The University of Texas MD Anderson Cancer Center, Division of Cancer Prevention and Population Sciences, Houston, TX, USA
| | - Ernest T Hawk
- The University of Texas MD Anderson Cancer Center, Division of Cancer Prevention and Population Sciences, Houston, TX, USA
| |
Collapse
|
14
|
Zhang X, Sun J, Xin W, Li Y, Ni L, Ma X, Zhang D, Zhang D, Zhang T, Du G. Anti-inflammation effect of methyl salicylate 2-O-β-D-lactoside on adjuvant induced-arthritis rats and lipopolysaccharide (LPS)-treated murine macrophages RAW264.7 cells. Int Immunopharmacol 2015; 25:88-95. [PMID: 25637446 DOI: 10.1016/j.intimp.2015.01.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 01/23/2015] [Accepted: 01/23/2015] [Indexed: 01/12/2023]
Abstract
Methyl salicylate 2-O-β-D-lactoside (MSL) is a derivative of natural salicylate isolated from Gaultheria yunnanensis (Franch.) Rehder, which is widely used for treating rheumatoid arthritis (RA), swelling and pain. The aim of the present study was to investigate the effect of MSL on the progression of adjuvant-induced arthritis (AIA) in rat in vivo and explore the anti-inflammatory effects and mechanism of MSL in lipopolysaccharide (LPS)-treated murine macrophages RAW264.7 cells in vitro. Our results showed that MSL significantly inhibited the arthritis progression in AIA rats, decreasing the right hind paw swelling and ankle diameter, attenuating histopathological changes and suppressing the plasma levels of TNF-α and IL-1β in AIA rats. Besides, MSL had potent anti-inflammatory effects on the LPS-activated RAW264.7. MSL dose-dependently inhibited the activity of COX-1, and COX-2. Moreover, MSL prominently inhibited LPS-induced activation of MAPK in RAW264.7 cells by blocking phosphorylation of p38 and ERK. Our study suggests that MSL may be effective in the treatment of inflammatory diseases by inhibiting the pro-inflammatory cytokine production and regulating the MAPK signal pathway.
Collapse
Affiliation(s)
- Xue Zhang
- Institute of Materia Medica, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100050, China.
| | - Jialin Sun
- Pharmacy Department of the Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Wenyu Xin
- Binzhou Medical University, Yantai 264003, China.
| | - Yongjie Li
- Institute of Materia Medica, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100050, China.
| | - Lin Ni
- Institute of Materia Medica, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Xiaowei Ma
- Institute of Materia Medica, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Dan Zhang
- Institute of Materia Medica, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Dongming Zhang
- Institute of Materia Medica, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Tiantai Zhang
- Institute of Materia Medica, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100050, China.
| | - Guanhua Du
- Institute of Materia Medica, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100050, China.
| |
Collapse
|
15
|
Francisella tularensis LVS induction of prostaglandin biosynthesis by infected macrophages requires specific host phospholipases and lipid phosphatases. Infect Immun 2014; 82:3299-311. [PMID: 24866789 DOI: 10.1128/iai.02060-14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Francisella tularensis induces the synthesis of prostaglandin E(2) (PGE(2)) by infected macrophages to alter host immune responses, thus providing a survival advantage to the bacterium. We previously demonstrated that PGE(2) synthesis by F. tularensis-infected macrophages requires cytosolic phospholipase A2 (cPLA(2)), cyclooxygenase 2 (COX-2), and microsomal prostaglandin E synthase 1 (mPGES1). During inducible PGE(2) synthesis, cPLA(2) hydrolyzes arachidonic acid (AA) from cellular phospholipids to be converted to PGE(2). However, in F. tularensis-infected macrophages we observed a temporal disconnect between Ser505-cPLA(2) phosphorylation (a marker of activation) and PGE(2) synthesis. These results suggested to us that cPLA(2) is not responsible for the liberation of AA to be converted into PGE(2) by F. tularensis-infected macrophages. Utilizing small-molecule inhibitors, we demonstrated that phospholipase D and diacylglycerol lipase were required for providing AA for PGE(2) biosynthesis. cPLA(2), on the other hand, was required for macrophage cytokine responses to F. tularensis. We also demonstrated for the first time that lipin-1 and PAP2a contribute to macrophage inflammation in response to F. tularensis. Our results identify both an alternative pathway for inducible PGE(2) synthesis and a role for lipid-modifying enzymes in the regulation of macrophage inflammatory function.
Collapse
|